Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
ACS Med Chem Lett ; 12(12): 1912-1919, 2021 Dec 09.
Article in English | MEDLINE | ID: mdl-34917254

ABSTRACT

The selective inhibition of RET kinase as a treatment for relevant cancer types including lung adenocarcinoma has garnered considerable interest in recent years and prompted a variety of efforts toward the discovery of small-molecule therapeutics. Hits uncovered via the analysis of archival kinase data ultimately led to the identification of a promising pyrrolo[2,3-d]pyrimidine scaffold. The optimization of this pyrrolo[2,3-d]pyrimidine core resulted in compound 1, which demonstrated potent in vitro RET kinase inhibition and robust in vivo efficacy in RET-driven tumor xenografts upon multiday dosing in mice. The administration of 1 was well-tolerated at established efficacious doses (10 and 30 mg/kg, po, qd), and plasma exposure levels indicated a minimal risk of KDR or hERG inhibition in vivo, as evaluated by Miles assay and free plasma concentrations, respectively.

2.
J Med Chem ; 64(8): 4857-4869, 2021 04 22.
Article in English | MEDLINE | ID: mdl-33821636

ABSTRACT

LONP1 is an AAA+ protease that maintains mitochondrial homeostasis by removing damaged or misfolded proteins. Elevated activity and expression of LONP1 promotes cancer cell proliferation and resistance to apoptosis-inducing reagents. Despite the importance of LONP1 in human biology and disease, very few LONP1 inhibitors have been described in the literature. Herein, we report the development of selective boronic acid-based LONP1 inhibitors using structure-based drug design as well as the first structures of human LONP1 bound to various inhibitors. Our efforts led to several nanomolar LONP1 inhibitors with little to no activity against the 20S proteasome that serve as tool compounds to investigate LONP1 biology.


Subject(s)
ATP-Dependent Proteases/antagonists & inhibitors , Drug Design , Mitochondrial Proteins/antagonists & inhibitors , Protease Inhibitors/chemistry , ATP-Dependent Proteases/metabolism , Binding Sites , Boronic Acids/chemistry , Boronic Acids/metabolism , Boronic Acids/pharmacology , Bortezomib/chemistry , Bortezomib/metabolism , Cell Line , Cell Survival/drug effects , Humans , Mitochondrial Proteins/metabolism , Molecular Docking Simulation , Protease Inhibitors/metabolism , Protease Inhibitors/pharmacology , Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/metabolism , Protein Subunits/antagonists & inhibitors , Protein Subunits/metabolism , Structure-Activity Relationship
3.
J Med Chem ; 63(19): 10773-10781, 2020 10 08.
Article in English | MEDLINE | ID: mdl-32667203

ABSTRACT

Visceral leishmaniasis is responsible for up to 30,000 deaths every year. Current treatments have shortcomings that include toxicity and variable efficacy across endemic regions. Previously, we reported the discovery of GNF6702, a selective inhibitor of the kinetoplastid proteasome, which cleared parasites in murine models of leishmaniasis, Chagas disease, and human African trypanosomiasis. Here, we describe the discovery and characterization of LXE408, a structurally related kinetoplastid-selective proteasome inhibitor currently in Phase 1 human clinical trials. Furthermore, we present high-resolution cryo-EM structures of the Leishmania tarentolae proteasome in complex with LXE408, which provides a compelling explanation for the noncompetitive mode of binding of this novel class of inhibitors of the kinetoplastid proteasome.


Subject(s)
Antiprotozoal Agents/chemistry , Antiprotozoal Agents/pharmacology , Leishmaniasis, Visceral/drug therapy , Oxazoles/chemistry , Oxazoles/pharmacology , Proteasome Inhibitors/chemistry , Proteasome Inhibitors/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Animals , Antiprotozoal Agents/therapeutic use , Dogs , Humans , Leishmania donovani/drug effects , Leishmania donovani/isolation & purification , Leishmania major/drug effects , Leishmania major/isolation & purification , Leishmaniasis, Visceral/parasitology , Liver/parasitology , Macaca fascicularis , Mice , Mice, Inbred BALB C , Oxazoles/therapeutic use , Proteasome Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Rats , Rats, Sprague-Dawley , Triazoles/chemistry
4.
ACS Med Chem Lett ; 11(4): 558-565, 2020 Apr 09.
Article in English | MEDLINE | ID: mdl-32292564

ABSTRACT

RET (REarranged during Transfection) kinase gain-of-function aberrancies have been identified as potential oncogenic drivers in lung adenocarcinoma, along with several other cancer types, prompting the discovery and assessment of selective inhibitors. Internal mining and analysis of relevant kinase data informed the decision to investigate a pyrazolo[1,5-a]pyrimidine scaffold, where subsequent optimization led to the identification of compound WF-47-JS03 (1), a potent RET kinase inhibitor with >500-fold selectivity against KDR (Kinase insert Domain Receptor) in cellular assays. In subsequent mouse in vivo studies, compound 1 demonstrated effective brain penetration and was found to induce strong regression of RET-driven tumor xenografts at a well-tolerated dose (10 mg/kg, po, qd). Higher doses of 1, however, were poorly tolerated in mice, similar to other pyrazolo[1,5-a]pyrimidine compounds at or near the efficacious dose, and indicative of the narrow therapeutic windows seen with this scaffold.

5.
Nat Methods ; 15(11): 941-946, 2018 11.
Article in English | MEDLINE | ID: mdl-30297964

ABSTRACT

CRISPR-Cas9 screening allows genome-wide interrogation of gene function. Currently, to achieve the high and uniform Cas9 expression desirable for screening, one needs to engineer stable and clonal Cas9-expressing cells-an approach that is not applicable in human primary cells. Guide Swap permits genome-scale pooled CRISPR-Cas9 screening in human primary cells by exploiting the unexpected finding that editing by lentivirally delivered, targeted guide RNAs (gRNAs) occurs efficiently when Cas9 is introduced in complex with nontargeting gRNA. We validated Guide Swap in depletion and enrichment screens in CD4+ T cells. Next, we implemented Guide Swap in a model of ex vivo hematopoiesis, and identified known and previously unknown regulators of CD34+ hematopoietic stem and progenitor cell (HSPC) expansion. We anticipate that this platform will be broadly applicable to other challenging cell types, and thus will enable discovery in previously inaccessible but biologically relevant human primary cell systems.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems , Gene Editing , Genome, Human , Hematopoietic Stem Cells/metabolism , RNA, Guide, Kinetoplastida/genetics , CD8-Positive T-Lymphocytes/cytology , Cells, Cultured , HEK293 Cells , Hematopoietic Stem Cells/cytology , Humans
6.
ACS Med Chem Lett ; 8(10): 1048-1053, 2017 Oct 12.
Article in English | MEDLINE | ID: mdl-29057049

ABSTRACT

NOD2 (nucleotide-binding oligomerization domain-containing protein 2) is an internal pattern recognition receptor that recognizes bacterial peptidoglycan and stimulates host immune responses. Dysfunction of NOD2 pathway has been associated with a number of autoinflammatory disorders. To date, direct inhibitors of NOD2 have not been described due to technical challenges of targeting the oligomeric protein complex. Receptor interacting protein kinase 2 (RIPK2) is an intracellular serine/threonine/tyrosine kinase, a key signaling partner, and an obligate kinase for NOD2. As such, RIPK2 represents an attractive target to probe the pathological roles of NOD2 pathway. To search for selective RIPK2 inhibitors, we employed virtual library screening (VLS) and structure based design that eventually led to a potent and selective RIPK2 inhibitor 8 with excellent oral bioavailability, which was used to evaluate the effects of inhibition of RIPK2 in various in vitro assays and ex vivo and in vivo pharmacodynamic models.

7.
Nature ; 537(7619): 229-233, 2016 09 08.
Article in English | MEDLINE | ID: mdl-27501246

ABSTRACT

Chagas disease, leishmaniasis and sleeping sickness affect 20 million people worldwide and lead to more than 50,000 deaths annually. The diseases are caused by infection with the kinetoplastid parasites Trypanosoma cruzi, Leishmania spp. and Trypanosoma brucei spp., respectively. These parasites have similar biology and genomic sequence, suggesting that all three diseases could be cured with drugs that modulate the activity of a conserved parasite target. However, no such molecular targets or broad spectrum drugs have been identified to date. Here we describe a selective inhibitor of the kinetoplastid proteasome (GNF6702) with unprecedented in vivo efficacy, which cleared parasites from mice in all three models of infection. GNF6702 inhibits the kinetoplastid proteasome through a non-competitive mechanism, does not inhibit the mammalian proteasome or growth of mammalian cells, and is well-tolerated in mice. Our data provide genetic and chemical validation of the parasite proteasome as a promising therapeutic target for treatment of kinetoplastid infections, and underscore the possibility of developing a single class of drugs for these neglected diseases.


Subject(s)
Chagas Disease/drug therapy , Kinetoplastida/drug effects , Kinetoplastida/enzymology , Leishmaniasis/drug therapy , Proteasome Endopeptidase Complex/drug effects , Proteasome Inhibitors/pharmacology , Proteasome Inhibitors/therapeutic use , Pyrimidines/pharmacology , Triazoles/pharmacology , Trypanosomiasis, African/drug therapy , Animals , Chagas Disease/parasitology , Chymotrypsin/antagonists & inhibitors , Chymotrypsin/metabolism , Disease Models, Animal , Female , Humans , Inhibitory Concentration 50 , Leishmaniasis/parasitology , Mice , Molecular Structure , Molecular Targeted Therapy , Proteasome Inhibitors/adverse effects , Proteasome Inhibitors/classification , Pyrimidines/adverse effects , Pyrimidines/chemistry , Pyrimidines/therapeutic use , Species Specificity , Triazoles/adverse effects , Triazoles/chemistry , Triazoles/therapeutic use , Trypanosomiasis, African/parasitology
8.
Biochem J ; 430(3): 425-37, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20632993

ABSTRACT

ALK (anaplastic lymphoma kinase) is an RTK (receptor tyrosine kinase) of the IRK (insulin receptor kinase) superfamily, which share an YXXXYY autophosphorylation motif within their A-loops (activation loops). A common activation and regulatory mechanism is believed to exist for members of this superfamily typified by IRK and IGF1RK (insulin-like growth factor receptor kinase-1). Chromosomal translocations involving ALK were first identified in anaplastic large-cell lymphoma, a subtype of non-Hodgkin's lymphoma, where aberrant fusion of the ALK kinase domain with the NPM (nucleophosmin) dimerization domain results in autophosphosphorylation and ligand-independent activation. Activating mutations within the full-length ALK kinase domain, most commonly R1275Q and F1174L, which play a major role in neuroblastoma, were recently identified. To provide a structural framework for understanding these mutations and to guide structure-assisted drug discovery efforts, the X-ray crystal structure of the unphosphorylated ALK catalytic domain was determined in the apo, ADP- and staurosporine-bound forms. The structures reveal a partially inactive protein kinase conformation distinct from, and lacking, many of the negative regulatory features observed in inactive IGF1RK/IRK structures in their unphosphorylated forms. The A-loop adopts an inhibitory pose where a short proximal A-loop helix (alphaAL) packs against the alphaC helix and a novel N-terminal beta-turn motif, whereas the distal portion obstructs part of the predicted peptide-binding region. The structure helps explain the reported unique peptide substrate specificity and the importance of phosphorylation of the first A-loop Tyr1278 for kinase activity and NPM-ALK transforming potential. A single amino acid difference in the ALK substrate peptide binding P-1 site (where the P-site is the phosphoacceptor site) was identified that, in conjunction with A-loop sequence variation including the RAS (Arg-Ala-Ser)-motif, rationalizes the difference in the A-loop tyrosine autophosphorylation preference between ALK and IGF1RK/IRK. Enzymatic analysis of recombinant R1275Q and F1174L ALK mutant catalytic domains confirms the enhanced activity and transforming potential of these mutants. The transforming ability of the full-length ALK mutants in soft agar colony growth assays corroborates these findings. The availability of a three-dimensional structure for ALK will facilitate future structure-function and rational drug design efforts targeting this receptor tyrosine kinase.


Subject(s)
Catalytic Domain , Mutant Proteins/chemistry , Protein Conformation , Protein-Tyrosine Kinases/chemistry , Adenosine Diphosphate/chemistry , Adenosine Diphosphate/metabolism , Amino Acid Motifs/genetics , Amino Acid Sequence , Anaplastic Lymphoma Kinase , Animals , Cell Line , Crystallization , Crystallography, X-Ray , Humans , Kinetics , Models, Molecular , Molecular Sequence Data , Mutagenesis , Mutant Proteins/metabolism , Neuroblastoma/enzymology , Neuroblastoma/genetics , Phosphorylation , Protein Binding , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Receptor Protein-Tyrosine Kinases , Sequence Homology, Amino Acid , Spodoptera , Staurosporine/chemistry , Staurosporine/metabolism , Substrate Specificity
9.
Glycobiology ; 17(8): 805-19, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17513886

ABSTRACT

Treatment options for androgen-independent prostate cancer cells are limited. Therefore, it is critical to identify agents that induce death of both androgen-responsive and androgen-insensitive cells. Here we demonstrate that a product of plant cell walls, pectin, is capable of inducing apoptosis in androgen-responsive (LNCaP) and androgen-independent (LNCaP C4-2) human prostate cancer cells. Commercially available fractionated pectin powder (FPP) induced apoptosis (approximately 40-fold above non-treated cells) in both cell lines as determined by the Apoptosense assay and activation of caspase-3 and its substrate, poly(ADP-ribose) polymerase. Conversely, citrus pectin (CP) and the pH-modified CP, PectaSol, had little or no apoptotic activity. Glycosyl residue composition and linkage analyses revealed no significant differences among the pectins. Mild base treatment to remove ester linkages destroyed FPP's apoptotic activity and yielded homogalacturonan (HG) oligosaccharides. The treatment of FPP with pectinmethylesterase to remove galacturonosyl carboxymethylesters and/or with endopolygalacturonase to cleave nonmethylesterified HG caused no major reduction in apoptotic activity, implicating the requirement for a base-sensitive linkage other than the carboxymethylester. Heat treatment of CP (HTCP) led to the induction of significant levels of apoptosis comparable to FPP, suggesting a means for generating apoptotic pectic structures. These results indicate that specific structural elements within pectin are responsible for the apoptotic activity, and that this structure can be generated, or enriched for, by heat treatment of CP. These findings provide the foundation for mechanistic studies of pectin apoptotic activity and a basis for the development of pectin-based pharmaceuticals, nutraceuticals, or recommended diet changes aimed at combating prostate cancer occurrence and progression.


Subject(s)
Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis , Pectins/chemistry , Pectins/pharmacology , Prostatic Neoplasms/drug therapy , Antineoplastic Agents, Phytogenic/therapeutic use , Cell Line, Tumor , Cells, Cultured , Endothelial Cells/drug effects , Humans , Hydrogen-Ion Concentration , Male , Oligosaccharides/metabolism , Pectins/therapeutic use , Prostatic Neoplasms/pathology , Protein Denaturation
10.
J Bacteriol ; 188(17): 6168-78, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16923883

ABSTRACT

Rhizobia are nitrogen-fixing bacteria that establish endosymbiotic associations with legumes. Nodule formation depends on various bacterial carbohydrates, including lipopolysaccharides, K-antigens, and exopolysaccharides (EPS). An acidic EPS from Rhizobium sp. strain NGR234 consists of glucosyl (Glc), galactosyl (Gal), glucuronosyl (GlcA), and 4,6-pyruvylated galactosyl (PvGal) residues with beta-1,3, beta-1,4, beta-1,6, alpha-1,3, and alpha-1,4 glycoside linkages. Here we examined the role of NGR234 genes in the synthesis of EPS. Deletions within the exoF, exoL, exoP, exoQ, and exoY genes suppressed accumulation of EPS in bacterial supernatants, a finding that was confirmed by chemical analyses. The data suggest that the repeating subunits of EPS are assembled by an ExoQ/ExoP/ExoF-dependent mechanism, which is related to the Wzy polymerization system of group 1 capsular polysaccharides in Escherichia coli. Mutation of exoK (NGROmegaexoK), which encodes a putative glycanase, resulted in the absence of low-molecular-weight forms of EPS. Analysis of the extracellular carbohydrates revealed that NGROmegaexoK is unable to accumulate exo-oligosaccharides (EOSs), which are O-acetylated nonasaccharide subunits of EPS having the formula Gal(Glc)5(GlcA)2PvGal. When used as inoculants, both the exo-deficient mutants and NGROmegaexoK were unable to form nitrogen-fixing nodules on some hosts (e.g., Albizia lebbeck and Leucaena leucocephala), but they were able to form nitrogen-fixing nodules on other hosts (e.g., Vigna unguiculata). EOSs of the parent strain were biologically active at very low levels (yield in culture supernatants, approximately 50 microg per liter). Thus, NGR234 produces symbiotically active EOSs by enzymatic degradation of EPS, using the extracellular endo-beta-1,4-glycanase encoded by exoK (glycoside hydrolase family 16). We propose that the derived EOSs (and not EPS) are bacterial components that play a crucial role in nodule formation in various legumes.


Subject(s)
Fabaceae/microbiology , Oligosaccharides/physiology , Rhizobium/physiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Genes, Bacterial , Genetic Complementation Test , Glycoside Hydrolases/genetics , Glycoside Hydrolases/metabolism , Mutation , Oligosaccharides/metabolism , Polysaccharides, Bacterial/metabolism , Rhizobium/genetics , Symbiosis
11.
J Bacteriol ; 186(19): 6617-25, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15375143

ABSTRACT

Analysis of two exopolysaccharide-deficient mutants of Rhizobium leguminosarum, RBL5808 and RBL5812, revealed independent Tn5 transposon integrations in a single gene, designated exo5. As judged from structural and functional homology, this gene encodes a UDP-glucose dehydrogenase responsible for the oxidation of UDP-glucose to UDP-glucuronic acid. A mutation in exo5 affects all glucuronic acid-containing polysaccharides and, consequently, all galacturonic acid-containing polysaccharides. Exo5-deficient rhizobia do not produce extracellular polysaccharide (EPS) or capsular polysaccharide (CPS), both of which contain glucuronic acid. Carbohydrate composition analysis and nuclear magnetic resonance studies demonstrated that EPS and CPS from the parent strain have very similar structures. Lipopolysaccharide (LPS) molecules produced by the mutant strains are deficient in galacturonic acid, which is normally present in the core and lipid A portions of the LPS. The sensitivity of exo5 mutant rhizobia to hydrophobic compounds shows the involvement of the galacturonic acid residues in the outer membrane structure. Nodulation studies with Vicia sativa subsp. nigra showed that exo5 mutant rhizobia are impaired in successful infection thread colonization. This is caused by strong agglutination of EPS-deficient bacteria in the root hair curl. Root infection could be restored by simultaneous inoculation with a Nod factor-defective strain which retained the ability to produce EPS and CPS. However, in this case colonization of the nodule tissue was impaired.


Subject(s)
Genes, Bacterial/physiology , Polysaccharides, Bacterial/biosynthesis , Rhizobium leguminosarum/genetics , Vicia sativa/microbiology , Bacterial Capsules/biosynthesis , Rhizobium leguminosarum/metabolism , Rhizobium leguminosarum/pathogenicity
SELECTION OF CITATIONS
SEARCH DETAIL
...